Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Nucleic Acids Res ; 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39011894

ABSTRACT

RNA helicases are involved in RNA metabolism in an ATP-dependent manner. Although many RNA helicases unwind the RNA structure and/or remove proteins from the RNA, some can load their interacting proteins onto RNAs. Here, we developed an in vitro strategy to identify the ATP-dependent factors involved in spliceosomal uridine-rich small nuclear RNA (U snRNA) export. We identified the RNA helicase UAP56/DDX39B, a component of the mRNA export complex named the transcription-export (TREX) complex, and its closely related RNA helicase URH49/DDX39A as the factors that stimulated RNA binding of PHAX, an adapter protein for U snRNA export. ALYREF, another TREX component, acted as a bridge between PHAX and UAP56/DDX39B. We also showed that UAP56/DDX39B and ALYREF participate in U snRNA export through a mechanism distinct from that of mRNA export. This study describes a novel aspect of the TREX components for U snRNP biogenesis and highlights the loading activity of RNA helicases.

2.
Nucleic Acids Res ; 51(3): 1393-1408, 2023 02 22.
Article in English | MEDLINE | ID: mdl-36620872

ABSTRACT

In eukaryotic cells, various classes of RNAs are exported to the cytoplasm by class-specific factors. Accumulating evidence has shown that export factors affect the fate of RNA, demonstrating the importance of proper RNA classification upon export. We previously reported that RNA polymerase II transcripts were classified after synthesis depending on their length, and identified heterogeneous nuclear ribonucleoprotein (hnRNP) C as the key classification factor. HnRNP C inhibits the recruitment of PHAX, an adapter protein for spliceosomal U snRNA export, to long transcripts, navigating these RNAs to the mRNA export pathway. However, the mechanisms by which hnRNP C inhibits PHAX recruitment to mRNA remain unknown. We showed that the cap-binding complex, a bridging factor between m7G-capped RNA and PHAX, directly interacted with hnRNP C on mRNA. Additionally, we revealed that the tetramer-forming activity of hnRNP C and its strong RNA-binding activity were crucial for the inhibition of PHAX binding to longer RNAs. These results suggest that mRNA is wrapped around the hnRNP C tetramer without a gap from the cap, thereby impeding the recruitment of PHAX. The results obtained on the mode of length-specific RNA classification by the hnRNP C tetramer will provide mechanistic insights into hnRNP C-mediated RNA biogenesis.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein Group C , RNA Polymerase II , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA Polymerase II/metabolism , RNA, Messenger/metabolism , RNA, Small Nuclear/genetics , Eukaryotic Cells/metabolism
3.
Genes Cells ; 26(1): 18-30, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33147372

ABSTRACT

Primary RNA transcripts are processed in a plethora of ways to become mature functional forms. In one example, human spliceosomal U snRNAs are matured at their 3'-end by an exonuclease termed TOE1. This process is important because mutations in TOE1 gene can cause a human genetic disease, pontocerebellar hypoplasia (PCH). Nevertheless, TOE1 may not be the only maturation exonuclease for U snRNAs in the cell. Here, we biochemically identify two exonucleolytic factors, Interferon-stimulated gene 20-kDa protein (ISG20) and the nuclear exosome as such candidates, using a newly developed in vitro system that recapitulates 3'-end maturation of U1 snRNA. However, extensive 3'-end sequencing of endogenous U1 snRNA of the knockdown (KD) cells revealed that these factors are not the maturation factors per se. Instead, the nascent transcripts of the spliceosomal U snRNAs as well as of unstable U1 variants were found to increase in quantity upon KD of the factors. These results indicated that ISG20 and the nuclear exosome promote the degradation of nascent spliceosomal U snRNAs and U1 variants, and therefore implied their role in the quality control of newly synthesized U snRNAs.


Subject(s)
Exoribonucleases/metabolism , Exosomes/metabolism , RNA, Small Nuclear/metabolism , Spliceosomes/metabolism , Cell Nucleus/metabolism , Exoribonucleases/genetics , HeLa Cells , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Stability , RNA, Small Nuclear/genetics
4.
RNA ; 26(11): 1716-1725, 2020 11.
Article in English | MEDLINE | ID: mdl-32759388

ABSTRACT

PHAX (phosphorylated adaptor for RNA export) promotes nuclear export of short transcripts of RNA polymerase II such as spliceosomal U snRNA precursors, as well as intranuclear transport of small nucleolar RNAs (snoRNAs). However, it remains unknown whether PHAX has other critical functions. Here we show that PHAX is required for efficient DNA damage response (DDR) via regulation of phosphorylated histone variant H2AX (γH2AX), a key factor for DDR. Knockdown of PHAX led to a significant reduction of H2AX mRNA levels, through inhibition of both transcription of the H2AX gene and nuclear export of H2AX mRNA, one of the shortest mRNAs in the cell. As a result, PHAX-knockdown cells become more sensitive to DNA damage due to a shortage of γH2AX. These results reveal a novel function of PHAX, which secures efficient DDR and hence genome stability.


Subject(s)
Histones/genetics , Histones/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Cell Line , DNA Damage , DNA Repair , Gene Expression , Gene Knockdown Techniques , Humans , Phosphorylation , Ultraviolet Rays/adverse effects
5.
Mol Cell Biol ; 40(4)2020 01 30.
Article in English | MEDLINE | ID: mdl-31818879

ABSTRACT

Nuclear paraspeckle assembly transcript 1 (NEAT1) is a long noncoding RNA that functions as an essential framework of subnuclear paraspeckle bodies. Of the two isoforms (NEAT1_1 and NEAT1_2) produced by alternative 3'-end RNA processing, the longer isoform, NEAT1_2, plays a crucial role in paraspeckle formation. Here, we demonstrate that the 3'-end processing and stability of NEAT1 RNAs are regulated by arsenic resistance protein 2 (ARS2), a factor interacting with the cap-binding complex (CBC) that binds to the m7G cap structure of RNA polymerase II transcripts. The knockdown of ARS2 inhibited the association between NEAT1 and mammalian cleavage factor I (CFIm), which produces the shorter isoform, NEAT1_1. Furthermore, the knockdown of ARS2 led to the preferential stabilization of NEAT1_2. As a result, NEAT1_2 RNA levels were markedly elevated in ARS2 knockdown cells, leading to an increase in the number of paraspeckles. These results reveal a suppressive role for ARS2 in NEAT1_2 expression and the subsequent formation of paraspeckles.


Subject(s)
Cell Nucleus/metabolism , Nuclear Proteins/genetics , RNA Processing, Post-Transcriptional/genetics , RNA, Long Noncoding/genetics , Cell Line, Tumor , Humans , RNA Interference , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics
6.
Stem Cell Reports ; 11(1): 228-241, 2018 07 10.
Article in English | MEDLINE | ID: mdl-30008325

ABSTRACT

The mechanistic/mammalian target of rapamycin complex 1 (mTORC1) regulates cellular function in various cell types. Although the role of mTORC1 in skeletogenesis has been investigated previously, here we show a critical role of mTORC1/4E-BPs/SOX9 axis in regulating skeletogenesis through its expression in undifferentiated mesenchymal cells. Inactivation of Raptor, a component of mTORC1, in limb buds before mesenchymal condensations resulted in a marked loss of both cartilage and bone. Mechanistically, we demonstrated that mTORC1 selectively controls the RNA translation of Sox9, which harbors a 5' terminal oligopyrimidine tract motif, via inhibition of the 4E-BPs. Indeed, introduction of Sox9 or a knockdown of 4E-BP1/2 in undifferentiated mesenchymal cells markedly rescued the deficiency of the condensation observed in Raptor-deficient mice. Furthermore, introduction of the Sox9 transgene rescued phenotypes of deficient skeletal growth in Raptor-deficient mice. These findings highlight a critical role of mTORC1 in mammalian skeletogenesis, at least in part, through translational control of Sox9 RNA.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Osteogenesis/genetics , Protein Biosynthesis , SOX9 Transcription Factor/genetics , Skeleton/metabolism , Animals , Cell Differentiation/genetics , Gene Expression , Mice , Mice, Transgenic , Phenotype , SOX9 Transcription Factor/metabolism , Skeleton/embryology
7.
Sci Rep ; 8(1): 1244, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29352143

ABSTRACT

Cellular translation should be precisely controlled in response to extracellular cues. However, knowledge is limited concerning signal transduction-regulated translation. In the present study, phosphorylation was identified in the 40S small subunit ribosomal protein uS7 (Yjr123w/previously called as Rps5) by Ypk1 and Pkc1, AGC family protein kinases in yeast Saccharomyces cerevisiae. Serine residue 223 (Ser223) of uS7 in the conserved C-terminal region was crucial for this phosphorylation event. S223A mutant uS7 caused severe reduction of small ribosomal subunit production, likely due to compromised interaction with Rio2, resulting in both reduced translation and reduced cellular proliferation. Contrary to optimal culture conditions, heat stressed S223A mutant cells exhibited increased heat resistance and induced heat shock proteins. Taken together, an intracellular signal transduction pathway involving Ypk1/Pkc1 seemed to play an important role in ribosome biogenesis and subsequent cellular translation, utilizing uS7 as a substrate.


Subject(s)
Protein Processing, Post-Translational , Ribosomal Proteins/metabolism , Ribosome Subunits, Small/metabolism , Heat-Shock Response , Mutation , Phosphorylation , Protein Domains , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction
8.
Front Mol Biosci ; 4: 93, 2017.
Article in English | MEDLINE | ID: mdl-29312956

ABSTRACT

RNA transport and regulated local translation play critically important roles in spatially restricting gene expression in neurons. Heterogeneous population of RNA granules serve as motile units to translocate, store, translate, and degrade mRNAs in the dendrites contain cis-elements and trans-acting factors such as RNA-binding proteins and microRNAs to convey stimulus-, transcript-specific local translation. Here we report a class of mRNA granules in human neuronal processes that are enriched in the nuclear cap-binding protein complex (CBC) and exon junction complex (EJC) core components, Y14 and eIF4AIII. These granules are physically associated with stabilized microtubules and are spatially segregated from eIF4E-enriched granules and P-bodies. The existence of mRNAs retaining both nuclear cap binding protein and EJC in the distal sites of neuronal processes suggests that some localized mRNAs have not yet undergone the "very first translation," which contribute to the spatio-temporal regulation of gene expression.

9.
Genes Cells ; 21(11): 1263-1269, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27659147

ABSTRACT

Localization of mRNA in neuronal cells is a critical process for spatiotemporal regulation of gene expression. Cytoplasmic localization of mRNA is often conferred by transport elements in 3' untranslated region (UTR). Activity-regulated cytoskeleton-associated protein (arc) mRNA is one of the localizing mRNAs in neuronal cells, and its localization is mediated by dendritic targeting element (DTE). As arc mRNA has introns in its 3' UTR, it was thought that arc mRNA is a natural target of nonsense-mediated mRNA decay (NMD). Here, we show that DTE in human arc 3' UTR has destabilizing activity of RNA independent of NMD pathway. DTE alone was able to cause instability of the reporter mRNA and this degradation was dependent on translation. Our results indicate that DTE has dual activity in mRNA transport and degradation, which suggests the novel spatiotemporal regulation mechanism of activity-dependent degradation of the mRNA.


Subject(s)
Cytoskeletal Proteins/genetics , Dendrites/metabolism , Nerve Tissue Proteins/genetics , Protein Sorting Signals , RNA Stability , RNA, Messenger/metabolism , 3' Untranslated Regions , Biological Transport , Cells, Cultured , Humans
11.
Genes Cells ; 20(4): 281-91, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25656399

ABSTRACT

The signal recognition particle is a ribonucleoprotein complex that is essential for the translocation of nascent proteins into the endoplasmic reticulum. It has been shown that the RNA component (SRP RNA) is exported from the nucleus by CRM1 in the budding yeast. However, how SRP RNA is exported in higher species has been elusive. Here, we show that SRP RNA does not use the CRM1 pathway in Xenopus oocytes. Instead, SRP RNA uses the same export pathway as pre-miRNA and tRNA as showed by cross-competition experiments. Consistently, the recombinant Exportin-5 protein specifically stimulated export of SRP RNA as well as of pre-miRNA and tRNA, whereas an antibody raised against Exportin-5 specifically inhibited export of the same RNA species. Moreover, biotinylated SRP RNA can pull down Exportin-5 but not CRM1 from HeLa cell nuclear extracts in a RanGTP-dependent manner. These results, taken together, strongly suggest that the principal export receptor for SRP RNA in vertebrates is Exportin-5 unlike in the budding yeast.


Subject(s)
Cell Nucleus/metabolism , Karyopherins/metabolism , RNA/metabolism , Signal Recognition Particle/metabolism , Vertebrates/metabolism , Active Transport, Cell Nucleus , Animals , HeLa Cells , Humans , MicroRNAs/metabolism , Microinjections/methods , Oocytes , RNA, Transfer/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Xenopus , Exportin 1 Protein
12.
Int J Mol Sci ; 16(2): 3705-21, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25671812

ABSTRACT

In eukaryotes, pre-mRNA splicing is an essential step for gene expression. We have been analyzing post-splicing intron turnover steps in higher eukaryotes. Here, we report protein interaction between human Debranching enzyme 1 (hDbr1) and several factors found in the Intron Large (IL) complex, which is an intermediate complex of the intron degradation pathway. The hDbr1 protein specifically interacts with xeroderma pigmentosum, complementeation group A (XPA)-binding protein 2 (Xab2). We also attempted to identify specific interactors of hDbr1. Co-immunoprecipitation experiments followed by mass spectrometry analysis identified a novel protein as one of the specific interactors of hDbr1. This protein is well conserved among many species and shows the highest similarity to yeast Drn1, so it is designated as human Dbr1 associated ribonuclease 1 (hDrn1). hDrn1 directly interacts with hDbr1 through protein-protein interaction. Furthermore, hDrn1 shuttles between the nucleus and the cytoplasm, as hDbr1 protein does. These findings suggest that hDrn1 has roles in both the nucleus and the cytoplasm, which are highly likely to involve hDbr1.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , RNA Nucleotidyltransferases/metabolism , Transcription Factors/metabolism , Animals , Cell Cycle Proteins/genetics , Coculture Techniques , HEK293 Cells , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Immunoprecipitation , Mass Spectrometry , Mice , NIH 3T3 Cells , Protein Interaction Maps , RNA Nucleotidyltransferases/genetics , RNA Splicing Factors , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
13.
Biochem Biophys Res Commun ; 457(1): 90-4, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25534857

ABSTRACT

Nonfunctional mutant ribosomal RNAs in 40S or 60S subunits are selectively degraded in eukaryotic cells (nonfunctional rRNA decay, NRD). We previously reported that NRD of 25S rRNA required cullin-E3 ligase Rtt101 and its associating factor Mms1, both of which are involved in DNA repair. Although Mms22, an accessory component of the E3 complex, was suggested to direct the E3 complex to DNA repair, the factor that directs the complex to 25S NRD currently remains unknown. We herein demonstrated that another accessory component, Crt10 was required for 25S NRD, but not for DNA repair, suggesting that this accessory component specifies the function of the E3 complex differently. We also identified two distinct Crt10-containing E3 complexes, one of which contained the Paf1 complex, a Pol-II binding complex that modulates the transcription of stress-related genes. Our results showed the convergence of multiple pathways for stresses that harm nucleic acids and provided a molecular framework for the substrate diversity of the E3 complex.


Subject(s)
Cullin Proteins/metabolism , DNA-Binding Proteins/metabolism , RNA Stability , RNA, Ribosomal/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , DNA Repair , Mutation , Protein Binding , Ribosomes/metabolism , Substrate Specificity , Ubiquitination
14.
Methods Cell Biol ; 122: 395-413, 2014.
Article in English | MEDLINE | ID: mdl-24857740

ABSTRACT

In eukaryotes, many RNA species are transcribed, processed in the nucleus, and exported to the cytoplasm, where they are destined to function or to be further matured. Some RNAs are even reimported to the nucleus. In addition, many RNAs are localized at specific nuclear bodies before their export and/or after their nuclear reimport. To understand how RNAs are transported, Xenopus oocytes are extremely useful cells, thanks to their large size. RNA transport can be easily examined by microinjecting radioactively or fluorescently labeled RNAs into Xenopus oocytes. Mammalian cultured cells are sometimes useful by virtue of RNA-FISH technique. Here, we describe methods to analyze RNA localization and export using these cells.


Subject(s)
Active Transport, Cell Nucleus/physiology , Cell Nucleus/metabolism , Oocytes/cytology , RNA Transport/physiology , Animals , Autoradiography/methods , Cell Line, Tumor , Digoxigenin/chemistry , Fluorescent Dyes , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , In Situ Hybridization, Fluorescence/methods , Microinjections , Phosphorus Radioisotopes , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , Staining and Labeling , Transcription, Genetic , Xenopus
15.
Nucleic Acids Res ; 42(10): 6645-58, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24753416

ABSTRACT

Nuclear RNA export pathways in eukaryotes are often linked to the fate of a given RNA. Therefore, the choice of export pathway should be well-controlled to avoid an unfavorable effect on gene expression. Although some RNAs could be exported by more than one pathway, little is known about how the choice is regulated. This issue is highlighted when the human immunodeficiency virus type 1 (HIV-1) Rev protein induces the export of singly spliced and unspliced HIV-1 transcripts. How these RNAs are exported is not well understood because such transcripts should have the possibility of utilizing CRM1-dependent export via Rev or cellular TAP/NXF1-dependent export via the transcription/export (TREX) complex, or both. Here we found that Rev suppressed TAP/NXF1-dependent export of model RNA substrates that recapitulated viral transcripts. In this effect, Rev interacted with the cap-binding complex and inhibited the recruitment of the TREX complex. Thus, Rev controls the identity of the factor occupying the cap-proximal region that determines the RNA export pathway. This ribonucleoprotein remodeling activity of Rev may favor viral gene expression.


Subject(s)
HIV-1/genetics , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , rev Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Drosophila Proteins/genetics , Fushi Tarazu Transcription Factors/genetics , HIV-1/metabolism , Karyopherins/metabolism , Nucleocytoplasmic Transport Proteins/antagonists & inhibitors , Oocytes/metabolism , RNA Cap-Binding Proteins/metabolism , RNA Splicing , RNA Transport , RNA, Viral/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Xenopus , Exportin 1 Protein
16.
Nucleic Acids Res ; 42(6): 3998-4007, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24413662

ABSTRACT

The assembly of spliceosomal U snRNPs in metazoans requires nuclear export of U snRNA precursors. Four factors, nuclear cap-binding complex (CBC), phosphorylated adaptor for RNA export (PHAX), the export receptor CRM1 and RanGTP, gather at the m(7)G-cap-proximal region and form the U snRNA export complex. Here we show that the multifunctional RNA-binding proteins p54nrb/NonO and PSF are U snRNA export stimulatory factors. These proteins, likely as a heterodimer, accelerate the recruitment of PHAX, and subsequently CRM1 and Ran onto the RNA substrates in vitro, which mediates efficient U snRNA export in vivo. Our results reveal a new layer of regulation for U snRNA export and, hence, spliceosomal U snRNP biogenesis.


Subject(s)
Cell Nucleus/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Octamer Transcription Factors/metabolism , RNA, Small Nuclear/metabolism , RNA-Binding Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Cytoplasm/metabolism , DNA-Binding Proteins , HeLa Cells , Humans , Karyopherins/metabolism , PTB-Associated Splicing Factor , Phosphoproteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Xenopus , ran GTP-Binding Protein/metabolism , Exportin 1 Protein
17.
Genes Cells ; 19(1): 78-87, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24304693

ABSTRACT

To identify the novel factors involved in the postsplicing intron turnover pathway, we carried out immunoprecipitation with known postsplicing factors, hPrp43 and TFIP11. As an interacting factor, we identified C2ORF3 protein by mass spectrometry. We found that C2ORF3 protein is present in the previously characterized Intron Large (IL) complex with an excised lariat intron. In vitro splicing using C2ORF3-depleted nuclear extracts showed significant repression of splicing, suggesting that C2ORF3 protein is required for pre-mRNA splicing through its presumable role in efficient intron turnover. Interestingly, C2ORF3 protein is localized in both the nucleoplasm and nucleoli, which suggests a potential function in rRNA processing.


Subject(s)
Introns , RNA Precursors/metabolism , Repressor Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Helicases/metabolism , RNA Precursors/genetics , RNA Splicing , RNA Splicing Factors , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism
18.
Sci Rep ; 3: 1090, 2013.
Article in English | MEDLINE | ID: mdl-23346348

ABSTRACT

In higher eukaryotes most genes contain multiple introns. Introns are excised from pre-mRNAs by splicing and eventually degraded in the nucleus. It is likely that rapid intron turnover in the nucleus is important in higher eukaryotes, but this pathway is poorly understood. In order to gain insights into this pathway, we analyzed the human lariat RNA debranching enzyme1 (hDbr1) protein that catalyzes debranching of lariat-intron RNAs. Transfection experiments demonstrate that hDbr1 is localized in a nucleoplasm of HeLa cells through a bipartite type nuclear localization signal near carboxyl-terminus. The conserved GNHE motif, originally identified in protein phosphatase protein family, is critical for hDbr1 to dissolve lariat structure in vitro. Furthermore, heterokaryon experiments show that hDbr1 is a nucleocytoplasmic shuttling protein, suggesting novel role(s) of hDbr1 in the cytoplasm.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Nuclear Localization Signals/metabolism , Phosphoprotein Phosphatases/metabolism , RNA Nucleotidyltransferases/genetics , RNA Nucleotidyltransferases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Cell Line, Tumor , Cell Nucleus/genetics , Cytoplasm/genetics , Drosophila Proteins , HEK293 Cells , HeLa Cells , Humans , Introns , Mice , Molecular Sequence Data , NIH 3T3 Cells , Nuclear Localization Signals/genetics , Phosphoprotein Phosphatases/genetics , RNA/genetics , RNA/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing , Sequence Alignment
19.
Translation (Austin) ; 1(2): e26402, 2013.
Article in English | MEDLINE | ID: mdl-26824023

ABSTRACT

In yeast Saccharomyces cerevisiae, 25S rRNA makes up the major mass and shape of the 60S ribosomal subunit. During translation initiation, the 60S subunit joins the 40S initiation complex, producing the 80S initiation complex. During elongation, the 60S subunit binds the CCA-ends of aminoacyl- and peptidyl-tRNAs at the A-loop and P-loop, respectively, transferring the peptide onto the α-amino group of the aminoacyl-tRNA. To study the role of 25S rRNA in translation in vivo, we randomly mutated 25S rRNA and isolated and characterized seven point mutations that affected yeast cell growth and polysome profiles. Four of these mutations, G651A, A1435U, A1446G and A1587G, change a base involved in base triples crucial for structural integrity. Three other mutations change bases near the ribosomal surface: C2879U and U2408C alter the A-loop and P-loop, respectively, and G1735A maps near a Eukarya-specific bridge to the 40S subunit. By polysome profiling in mmslΔ mutants defective in nonfunctional 25S rRNA decay, we show that some of these mutations are defective in both the initiation and elongation phases of translation. Of the mutants characterized, C2879U displays the strongest defect in translation initiation. The ribosome transit-time assay directly shows that this mutation is also defective in peptide elongation/termination. Thus, our genetic analysis not only identifies bases critical for structural integrity of the 60S subunit, but also suggests a role for bases near the peptidyl transferase center in translation initiation.

20.
RNA Biol ; 9(12): 1413-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23187719

ABSTRACT

In eukaryotic cells, many RNA species are exported from the nucleus to the cytoplasms. Different RNA species form distinct ribonucleoprotein (RNP) complexes for export, indicating specific RNA recognition by export proteins. Specific RNA recognition is usually achieved by specific RNA sequences or structures, but we have recently reported a molecular mechanism by which the formation of export RNP complexes is specified by RNA length. ( 1) RNA polymerase II (Pol II) synthesizes not only mRNAs but also shorter RNAs, including spliceosomal U snRNAs. Although the key U snRNA export factor, PHAX, can bind to mRNA in vitro, PHAX is excluded from mRNA in vivo. The heterotetramer of the heterogeneous nuclear RNP (hnRNP) C1/C2 specifically binds Pol II transcripts longer than 200-300 nt, and funnels them into the mRNA export pathway by inhibiting their binding by PHAX, whereas shorter transcripts not bound by the heterotetramer are committed to the U snRNA export pathway. Although this finding reveals a novel function of the C1/C2 heterotetramer and highlights the biological importance of RNA recognition by length, it has raised a number of new questions, some of which will be discussed in this article, together with some historical background of this finding.


Subject(s)
RNA Transport , RNA, Small Nuclear/metabolism , Transcription, Genetic , Animals , Base Sequence , Cell Nucleus/genetics , Cell Nucleus/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group C/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Histones/genetics , Histones/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Nucleic Acid Conformation , Protein Binding , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Nuclear/genetics , Spliceosomes/genetics , Spliceosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL